1
|
Wang Y, Fasching L, Wu F, Suvakov M, Huttner A, Berretta S, Roberts R, Leckman JF, Fernandez TV, Abyzov A, Vaccarino FM. Interneuron loss and microglia activation by transcriptome analyses in the basal ganglia of Tourette disorder. Biol Psychiatry 2025:S0006-3223(25)00064-2. [PMID: 39892689 DOI: 10.1016/j.biopsych.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/28/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Tourette disorder is characterized by motor hyperactivity and tics that are believed to originate in basal ganglia. Postmortem immunocytochemical analyses previously revealed decreases in cholinergic, parvalbumin, and somatostatin interneurons (IN) within the caudate/putamen of individuals with TS. METHODS We obtained transcriptome and open chromatin datasets by snRNAseq and snATAC-seq, respectively, from caudate/putamen postmortem specimens of 6 adult TS and 6 matched normal control (NC). Differential gene expression and differential chromatin accessibility analyses were performed in identified cell types. RESULTS The data reproduced the known cellular composition of the human striatum, including a majority of medium spiny neurons (MSN) and small populations of GABAergic and cholinergic IN. IN were decreased by ∼50% in TS brains, with no difference in other cell types. Differential gene expression analysis suggested that mitochondrial oxidative metabolism in MSN and synaptic adhesion and function in IN were both decreased in TS subjects, while there was activation of immune response in microglia. Gene expression changes correlated with changes in activity of cis-regulatory elements, suggesting a relationship of transcriptomic and regulatory abnormalities in MSN, OL and AST of TS brains. CONCLUSIONS This initial analysis of the TS basal ganglia transcriptome at the single cell level confirms the loss and synaptic dysfunction of basal ganglia IN, consistent with in vivo basal ganglia hyperactivity. In parallel, oxidative metabolism was decreased in MSN and correlated with activation of microglia cells, attributable at least in part to dysregulated activity of putative enhancers, implicating altered epigenomic regulation in TS.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Quantitative Health Sciences, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Liana Fasching
- Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Feinan Wu
- Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Milovan Suvakov
- Department of Quantitative Health Sciences, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Sabina Berretta
- McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Rosalinda Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - James F Leckman
- Child Study Center, Yale University, New Haven, CT 06520, USA
| | | | - Alexej Abyzov
- Department of Quantitative Health Sciences, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Flora M Vaccarino
- Child Study Center, Yale University, New Haven, CT 06520, USA; Department of Neuroscience, Yale University, New Haven, CT 06520, USA; Yale Kavli Institute for Neuroscience, New Haven, CT 06520, USA.
| |
Collapse
|
2
|
Dienel SJ, Wade KL, Fish KN, Lewis DA. Alterations in Prefrontal Cortical Somatostatin Neurons in Schizophrenia: Evidence for Weaker Inhibition of Pyramidal Neuron Dendrites. Biol Psychiatry 2025:S0006-3223(25)00052-6. [PMID: 39848397 DOI: 10.1016/j.biopsych.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Certain cognitive processes require inhibition provided by the somatostatin (SST) class of gamma-aminobutyric acid (GABA) neurons in the dorsolateral prefrontal cortex (DLPFC). This inhibition onto pyramidal neuron dendrites depends on both SST and GABA signaling. Although SST mRNA levels are lower in the DLPFC in schizophrenia, it is not known if SST neurons exhibit alterations in the capacity to synthesize GABA, principally via the 67-kilodalton isoform of glutamic acid decarboxylase (GAD67). METHODS GAD67 and SST mRNA levels were quantified in individual SST neurons using fluorescence in situ hybridization in DLPFC layers 2-superficial 3, where SST neurons are enriched, in schizophrenia (n=46) and unaffected comparison (n=46) individuals. Findings were compared to GAD67 and SST mRNA levels quantified by polymerase chain reaction and to final educational attainment, a proxy measure for cognitive functioning. RESULTS GAD67 (F1,84=13.1, p=0.0005, Cohen's d = -0.78) and SST (F1,84=10.1, p=0.002, Cohen's d = -0.64) mRNA levels in SST neurons were lower in schizophrenia, with no group differences in the relative density of SST neurons (F1,84=0.21, p=0.65). A presynaptic index of dendritic inhibition, derived by summing the alterations in GAD67 and SST mRNAs, was lower in 80.4% of individuals with schizophrenia and was associated with final educational attainment (adjusted odds ratio=1.44, p=0.022). CONCLUSIONS Deficits in both GAD67 and SST mRNAs within SST neurons indicate that these neurons have a markedly reduced ability to inhibit postsynaptic pyramidal neuron dendrites in schizophrenia. These alterations likely contribute to cognitive dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Samuel J Dienel
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh; Center for the Neural Basis of Cognition, Carnegie Mellon University
| | - Kirsten L Wade
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh
| | - Kenneth N Fish
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh; Center for the Neural Basis of Cognition, Carnegie Mellon University.
| |
Collapse
|
3
|
Cao W, Li H, Luo J. Prefrontal cortical circuits in social behaviors: an overview. J Zhejiang Univ Sci B 2024; 25:941-955. [PMID: 39626878 PMCID: PMC11634449 DOI: 10.1631/jzus.b2300743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/02/2024] [Indexed: 12/13/2024]
Abstract
Social behaviors are fundamental and intricate functions in both humans and animals, governed by the interplay of social cognition and emotions. A noteworthy feature of several neuropsychiatric disorders, including autism spectrum disorder (ASD) and schizophrenia (SCZ), is a pronounced deficit in social functioning. Despite a burgeoning body of research on social behaviors, the precise neural circuit mechanisms underpinning these phenomena remain to be elucidated. In this paper, we review the pivotal role of the prefrontal cortex (PFC) in modulating social behaviors, as well as its functional alteration in social disorders in ASD or SCZ. We posit that PFC dysfunction may represent a critical hub in the pathogenesis of psychiatric disorders characterized by shared social deficits. Furthermore, we delve into the intricate connectivity of the medial PFC (mPFC) with other cortical areas and subcortical brain regions in rodents, which exerts a profound influence on social behaviors. Notably, a substantial body of evidence underscores the role of N-methyl-D-aspartate receptors (NMDARs) and the proper functioning of parvalbumin-positive interneurons within the mPFC for social regulation. Our overarching goal is to furnish a comprehensive understanding of these intricate circuits and thereby contribute to the enhancement of both research endeavors and clinical practices concerning social behavior deficits.
Collapse
Affiliation(s)
- Wei Cao
- Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Zhejiang Philosophy and Social Science Laboratory for Research in Early Development and Childcare, Hangzhou Normal University, Hangzhou 311121, China
| | - Huiyi Li
- Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Jianhong Luo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310013, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
4
|
Couch ACM, Brown AM, Raimundo C, Solomon S, Taylor M, Sichlinger L, Matuleviciute R, Srivastava DP, Vernon AC. Transcriptional and cellular response of hiPSC-derived microglia-neural progenitor co-cultures exposed to IL-6. Brain Behav Immun 2024; 122:27-43. [PMID: 39098436 DOI: 10.1016/j.bbi.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
Elevated interleukin (IL-)6 levels during prenatal development have been linked to increased risk for neurodevelopmental disorders (NDD) in the offspring, but the mechanism remains unclear. Human-induced pluripotent stem cell (hiPSC) models offer a valuable tool to study the effects of IL-6 on features relevant for human neurodevelopment in vitro. We previously reported that hiPSC-derived microglia-like cells (MGLs) respond to IL-6, but neural progenitor cells (NPCs) in monoculture do not. Therefore, we investigated whether co-culturing hiPSC-derived MGLs with NPCs would trigger a cellular response to IL-6 stimulation via secreted factors from the MGLs. Using N=4 donor lines without psychiatric diagnosis, we first confirmed that NPCs can respond to IL-6 through trans-signalling when recombinant IL-6Ra is present, and that this response is dose-dependent. MGLs secreted soluble IL-6R, but at lower levels than found in vivo and below that needed to activate trans-signalling in NPCs. Whilst transcriptomic and secretome analysis confirmed that MGLs undergo substantial transcriptomic changes after IL-6 exposure and subsequently secrete a cytokine milieu, NPCs in co-culture with MGLs exhibited a minimal transcriptional response. Furthermore, there were no significant cell fate-acquisition changes when differentiated into post-mitotic cultures, nor alterations in synaptic densities in mature neurons. These findings highlight the need to investigate if trans-IL-6 signalling to NPCs is a relevant disease mechanism linking prenatal IL-6 exposure to increased risk for psychiatric disorders. Moreover, our findings underscore the importance of establishing more complex in vitro human models with diverse cell types, which may show cell-specific responses to microglia-released cytokines to fully understand how IL-6 exposure may influence human neurodevelopment.
Collapse
Affiliation(s)
- Amalie C M Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Amelia M Brown
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Catarina Raimundo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shiden Solomon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Morgan Taylor
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Laura Sichlinger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Rugile Matuleviciute
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
5
|
Okuda T, Kimoto S, Kawabata R, Bian Y, Tsubomoto M, Okamura K, Enwright JF, Kikuchi M, Lewis DA, Hashimoto T. Alterations in inhibitory neuron subtype-selective transcripts in the prefrontal cortex: comparisons across schizophrenia and mood disorders. Psychol Med 2024; 54:1-10. [PMID: 39478366 PMCID: PMC11578916 DOI: 10.1017/s0033291724002344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/19/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND In schizophrenia (SZ), impairments in cognitive functions, such as working memory, have been associated with alterations in certain types of inhibitory neurons that utilize the neurotransmitter γ-aminobutyric acid (GABA) in the dorsolateral prefrontal cortex (DLPFC). For example, GABA neurons that express parvalbumin (PV) or somatostatin (SST) have more prominent gene expression alterations than those that express vasoactive intestinal peptide (VIP). In bipolar disorder (BD) and major depression (MD), which exhibit similar, but less severe, cognitive impairments than SZ, alterations of transcript levels in GABA neurons have also been reported. However, the extent to which GABA neuron subtype-selective transcripts in the DLPFC are affected, and the relative magnitudes of the diagnosis-associated effects, have not been directly compared across SZ, BD, and MD in the same study. METHODS We used quantitative polymerase chain reaction to examine levels of GABA neuron subtype-selective transcripts (PV, potassium voltage-gated channel modifier subfamily-S member-3, SST, VIP, and calretinin mRNAs), as well as the pan-GABA neuron marker 67 kDa glutamate decarboxylase mRNA, in DLPFC total gray matter of 160 individuals, including those with SZ, BD, or MD and unaffected comparison (UC) individuals. RESULTS Relative to UC individuals, individuals with SZ exhibited large deficits in levels of all transcripts except for calretinin mRNA, whereas individuals with BD or MD showed a marked deficit only for PV or SST mRNAs, respectively. CONCLUSIONS These findings suggest that broader and more severe alterations in DLPFC GABA neurons might contribute to the greater cognitive impairments in SZ relative to BD and MD.
Collapse
Affiliation(s)
- Takeshi Okuda
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Kashihara, 634-8521, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yufan Bian
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Makoto Tsubomoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Kazuya Okamura
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, 641-8509, Japan
| | - John F. Enwright
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Research Center for Child Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Takanori Hashimoto
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry, National Hospital Organization Hokuriku Hospital, Nanto, 939-1893, Japan
| |
Collapse
|
6
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
7
|
Uliana DL, Lisboa JRF, Gomes FV, Grace AA. The excitatory-inhibitory balance as a target for the development of novel drugs to treat schizophrenia. Biochem Pharmacol 2024; 228:116298. [PMID: 38782077 PMCID: PMC11410545 DOI: 10.1016/j.bcp.2024.116298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
The intricate balance between excitation and inhibition (E/I) in the brain plays a crucial role in normative information processing. Dysfunctions in the E/I balance have been implicated in various psychiatric disorders, including schizophrenia (SCZ). In particular, abnormalities in GABAergic signaling, specifically in parvalbumin (PV)-containing interneurons, have been consistently observed in SCZ pathophysiology. PV interneuron function is vital for maintaining an ideal E/I balance, and alterations in PV interneuron-mediated inhibition contribute to circuit deficits observed in SCZ, including hippocampus hyperactivity and midbrain dopamine system overdrive. While current antipsychotic medications primarily target D2 dopamine receptors and are effective primarily in treating positive symptoms, novel therapeutic strategies aiming to restore the E/I balance could potentially mitigate not only positive symptoms but also negative symptoms and cognitive deficits. This could involve, for instance, increasing the inhibitory drive onto excitatory neurons or decreasing the putative enhanced pyramidal neuron activity due to functional loss of PV interneurons. Compounds targeting the glycine site at glutamate NMDA receptors and muscarinic acetylcholine receptors on PV interneurons that can increase PV interneuron drive, as well as drugs that increase the postsynaptic action of GABA, such as positive allosteric modulators of α5-GABA-A receptors, and decrease glutamatergic output, such as mGluR2/3 agonists, represent promising approaches. Preventive strategies aiming at E/I balance also represent a path to reduce the risk of transitioning to SCZ in high-risk individuals. Therefore, compounds with novel mechanisms targeting E/I balance provide optimism for more effective and tailored interventions in the management of SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joao Roberto F Lisboa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Hughes H, Brady LJ, Schoonover KE. GABAergic dysfunction in postmortem dorsolateral prefrontal cortex: implications for cognitive deficits in schizophrenia and affective disorders. Front Cell Neurosci 2024; 18:1440834. [PMID: 39381500 PMCID: PMC11458443 DOI: 10.3389/fncel.2024.1440834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
The microcircuitry within superficial layers of the dorsolateral prefrontal cortex (DLPFC), composed of excitatory pyramidal neurons and inhibitory GABAergic interneurons, has been suggested as the neural substrate of working memory performance. In schizophrenia, working memory impairments are thought to result from alterations of microcircuitry within the DLPFC. GABAergic interneurons, in particular, are crucially involved in synchronizing neural activity at gamma frequency, the power of which increases with working memory load. Alterations of GABAergic interneurons, particularly parvalbumin (PV) and somatostatin (SST) subtypes, are frequently observed in schizophrenia. Abnormalities of GABAergic neurotransmission, such as deficiencies in the 67 kDA isoform of GABA synthesis enzyme (GAD67), vesicular GABA transporter (vGAT), and GABA reuptake transporter 1 (GAT1) in presynaptic boutons, as well as postsynaptic alterations in GABA A receptor subunits further contribute to impaired inhibition. This review explores GABAergic abnormalities of the postmortem DLPFC in schizophrenia, with a focus on the roles of interneuron subtypes involved in cognition, and GABAergic neurotransmission within presynaptic boutons and postsynaptic alterations. Where available, comparisons between schizophrenia and affective disorders that share cognitive pathology such as bipolar disorder and major depressive disorder will be made. Challenges in directly measuring GABA levels are addressed, emphasizing the need for innovative techniques. Understanding GABAergic abnormalities and their implications for neural circuit dysfunction in schizophrenia is crucial for developing targeted therapies.
Collapse
Affiliation(s)
- Hannah Hughes
- Graduate Biomedical Sciences Program, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
| | - Lillian J. Brady
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Tuskegee, AL, United States
| | - Kirsten E. Schoonover
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Tuskegee, AL, United States
- Department of Psychology and Sociology, College of Arts and Sciences, Tuskegee University, Tuskegee, AL, United States
| |
Collapse
|
9
|
Cale JA, Chauhan EJ, Cleaver JJ, Fusciardi AR, McCann S, Waters HC, Žavbi J, King MV. GABAergic and inflammatory changes in the frontal cortex following neonatal PCP plus isolation rearing, as a dual-hit neurodevelopmental model for schizophrenia. Mol Neurobiol 2024; 61:6968-6983. [PMID: 38363536 PMCID: PMC11339149 DOI: 10.1007/s12035-024-03987-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/24/2024] [Indexed: 02/17/2024]
Abstract
The pathogenesis of schizophrenia begins in early neurodevelopment and leads to excitatory-inhibitory imbalance. It is therefore essential that preclinical models used to understand disease, select drug targets and evaluate novel therapeutics encompass similar neurochemical deficits. One approach to improved preclinical modelling incorporates dual-hit neurodevelopmental insults, like neonatal administration of phencyclidine (PCP, to disrupt development of glutamatergic circuitry) then post-weaning isolation (Iso, to mimic adolescent social stress). We recently showed that male Lister-hooded rats exposed to PCP-Iso exhibit reduced hippocampal expression of the GABA interneuron marker calbindin. The current study expanded on this by investigating changes to additional populations of GABAergic interneurons in frontal cortical and hippocampal tissue from the same animals (by immunohistochemistry) as well as levels of GABA itself (via ELISA). Because inflammatory changes are also implicated in schizophrenia, we performed additional immunohistochemical evaluations of Iba-1 positive microglia as well as ELISA analysis of IL-6 in the same brain regions. Single-hit isolation-reared and dual-hit PCP-Iso rats both showed reduced parvalbumin immunoreactivity in the prelimbic/infralimbic region of the frontal cortex. However, this was more widespread in PCP-Iso, extending to the medial/ventral and lateral/dorsolateral orbitofrontal cortices. Loss of GABAergic markers was accompanied by increased microglial activation in the medial/ventral orbitofrontal cortices of PCP-Iso, together with frontal cortical IL-6 elevations not seen following single-hit isolation rearing. These findings enhance the face validity of PCP-Iso, and we advocate the use of this preclinical model for future evaluation of novel therapeutics-especially those designed to normalise excitatory-inhibitory imbalance or reduce neuroinflammation.
Collapse
Affiliation(s)
- Jennifer A Cale
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Ethan J Chauhan
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Joshua J Cleaver
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Anthoio R Fusciardi
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Sophie McCann
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Hannah C Waters
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Juš Žavbi
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, The University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| |
Collapse
|
10
|
Gillespie B, Dunn A, Sundram S, Hill RA. Investigating 7,8-Dihydroxyflavone to combat maternal immune activation effects on offspring gene expression and behaviour. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111078. [PMID: 38950841 DOI: 10.1016/j.pnpbp.2024.111078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Infection during pregnancy is a substantial risk factor for the unborn child to develop autism or schizophrenia later in life, and is thought to be driven by maternal immune activation (MIA). MIA can be modelled by exposing pregnant mice to Polyinosinic: polycytidylic acid (Poly-I:C), a viral mimetic that induces an immune response and recapitulates in the offspring many neurochemical features of ASD and schizophrenia, including altered BDNF-TrkB signalling and disruptions to excitatory/inhibitory balance. Therefore, we hypothesised that a BDNF mimetic, 7,8-Dihydroxyflavone (7,8-DHF), administered prophylactically to the dam may prevent the neurobehavioural sequelae of disruptions induced by MIA. Dams were treated with 7,8-DHF in the drinking water (0.08 mg/ML) from gestational day (GD) 9-20 and were exposed to Poly-I:C at GD17 (20 mg/kg, i.p.). Foetal brains were collected 6 h post Poly-I:C exposure for RT-qPCR analysis of BDNF, cytokine, GABAergic and glutamatergic gene targets. A second adult cohort were tested in a battery of behavioural tests relevant to schizophrenia and the prefrontal cortex and ventral hippocampus dissected for RT-qPCR analysis. Foetal brains exposed to Poly-I:C showed increased IL-6, but reduced expression of Ntrk2 and multiple GABAergic and glutamatergic markers. Anxiety-like behaviour was observed in adult offspring prenatally exposed to poly-I:C, which was accompanied by altered expression of Gria2 in the prefrontal cortex and Gria4 in the ventral hippocampus. While 7-8 DHF normalised the expression of some glutamatergic (Grm5) and GABAergic (Gabra1) genes in Poly-I:C exposed offspring, it also led to substantial alterations in offspring not exposed to Poly-I:C. Furthermore, mice exposed to 7,8-DHF prenatally showed increased pre-pulse inhibition and reduced working memory in adulthood. These data advance understanding of how 7,8-DHF and MIA prenatal exposure impacts genes critical to excitatory/inhibitory pathways and related behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Ariel Dunn
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
11
|
Cameron D, Vinh NN, Prapaiwongs P, Perry EA, Walters JTR, Li M, O’Donovan MC, Bray NJ. Genetic Implication of Prenatal GABAergic and Cholinergic Neuron Development in Susceptibility to Schizophrenia. Schizophr Bull 2024; 50:1171-1184. [PMID: 38869145 PMCID: PMC11349020 DOI: 10.1093/schbul/sbae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
BACKGROUND The ganglionic eminences (GE) are fetal-specific structures that give rise to gamma-aminobutyric acid (GABA)- and acetylcholine-releasing neurons of the forebrain. Given the evidence for GABAergic, cholinergic, and neurodevelopmental disturbances in schizophrenia, we tested the potential involvement of GE neuron development in mediating genetic risk for the condition. STUDY DESIGN We combined data from a recent large-scale genome-wide association study of schizophrenia with single-cell RNA sequencing data from the human GE to test the enrichment of schizophrenia risk variation in genes with high expression specificity for developing GE cell populations. We additionally performed the single nuclei Assay for Transposase-Accessible Chromatin with Sequencing (snATAC-Seq) to map potential regulatory genomic regions operating in individual cell populations of the human GE, using these to test for enrichment of schizophrenia common genetic variant liability and to functionally annotate non-coding variants-associated with the disorder. STUDY RESULTS Schizophrenia common variant liability was enriched in genes with high expression specificity for developing neuron populations that are predicted to form dopamine D1 and D2 receptor-expressing GABAergic medium spiny neurons of the striatum, cortical somatostatin-positive GABAergic interneurons, calretinin-positive GABAergic neurons, and cholinergic neurons. Consistent with these findings, schizophrenia genetic risk was concentrated in predicted regulatory genomic sequence mapped in developing neuronal populations of the GE. CONCLUSIONS Our study implicates prenatal development of specific populations of GABAergic and cholinergic neurons in later susceptibility to schizophrenia, and provides a map of predicted regulatory genomic elements operating in cells of the GE.
Collapse
Affiliation(s)
- Darren Cameron
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Ngoc-Nga Vinh
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Parinda Prapaiwongs
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| | - Elizabeth A Perry
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - James T R Walters
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Meng Li
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| | - Michael C O’Donovan
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Nicholas J Bray
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
12
|
Han S. Bayesian Rare Variant Analysis Identifies Novel Schizophrenia Putative Risk Genes. J Pers Med 2024; 14:822. [PMID: 39202013 PMCID: PMC11355493 DOI: 10.3390/jpm14080822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
The genetics of schizophrenia is so complex that it involves both common variants and rare variants. Rare variant association studies of schizophrenia are challenging because statistical methods for rare variant analysis are under-powered due to the rarity of rare variants. The recent Schizophrenia Exome meta-analysis (SCHEMA) consortium, the largest consortium in this field to date, has successfully identified 10 schizophrenia risk genes from ultra-rare variants by burden test, while more risk genes remain to be discovered by more powerful rare variant association test methods. In this study, we use a recently developed Bayesian rare variant association method that is powerful for detecting sparse rare risk variants that implicates 88 new candidate risk genes associated with schizophrenia from the SCHEMA case-control sample. These newly identified genes are significantly enriched in autism risk genes and GO enrichment analysis indicates that new candidate risk genes are involved in mechanosensory behavior, regulation of cell size, neuron projection morphogenesis, and plasma-membrane-bounded cell projection morphogenesis, that may provide new insights on the etiology of schizophrenia.
Collapse
Affiliation(s)
- Shengtong Han
- School of Dentistry, Marquette University, Milwaukee, WI 53201-1881, USA
| |
Collapse
|
13
|
He H, Long J, Song X, Li Q, Niu L, Peng L, Wei X, Zhang R. A connectome-wide association study of altered functional connectivity in schizophrenia based on resting-state fMRI. Schizophr Res 2024; 270:202-211. [PMID: 38924938 DOI: 10.1016/j.schres.2024.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/09/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Aberrant resting-state functional connectivity is a neuropathological feature of schizophrenia (SCZ). Prior investigations into functional connectivity abnormalities have primarily employed seed-based connectivity analysis, necessitating predefined seed locations. To address this limitation, a data-driven multivariate method known as connectome-wide association study (CWAS) has been proposed for exploring whole-brain functional connectivity. METHODS We conducted a CWAS analysis involving 46 patients with SCZ and 40 age- and sex-matched healthy controls. Multivariate distance matrix regression (MDMR) was utilized to identify key nodes in the brain. Subsequently, we conducted a follow-up seed-based connectivity analysis to elucidate specific connectivity patterns between regions of interest (ROIs). Additionally, we explored the spatial correlation between changes in functional connectivity and underlying molecular architectures by examining correlations between neurotransmitter/transporter distribution densities and functional connectivity. RESULTS MDMR revealed the right medial frontal gyrus and the left calcarine sulcus as two key nodes. Follow-up analysis unveiled hypoconnectivity between the right medial frontal superior gyrus and the right fusiform gyrus, as well as hypoconnectivity between the left calcarine sulcus and the right lingual gyrus in SCZ. Notably, a significant association between functional connectivity strength and positive symptom severity was identified. Furthermore, altered functional connectivity patterns suggested potential dysfunctions in the dopamine, serotonin, and gamma-aminobutyric acid systems. CONCLUSIONS This study elucidated reduced functional connectivity both within and between the medial frontal regions and the occipital cortex in patients with SCZ. Moreover, it indicated potential alterations in molecular architecture, thereby expanding current knowledge regarding neurobiological changes associated with SCZ.
Collapse
Affiliation(s)
- Huawei He
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jixin Long
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoqi Song
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qian Li
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lijing Niu
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lanxin Peng
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First Affiliated Hospital, Guangzhou, China.
| | - Ruibin Zhang
- Cognitive Control and Brain Healthy Laboratory, Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China; Department of Psychiatry, Zhujiang Hospital, Southern Medical University, Guangzhou, PRC, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for PsychiatricDisorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, PR China.
| |
Collapse
|
14
|
Schoonover KE, Dienel SJ, Holly Bazmi H, Enwright JF, Lewis DA. Altered excitatory and inhibitory ionotropic receptor subunit expression in the cortical visuospatial working memory network in schizophrenia. Neuropsychopharmacology 2024; 49:1183-1192. [PMID: 38548877 PMCID: PMC11109337 DOI: 10.1038/s41386-024-01854-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/14/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024]
Abstract
Dysfunction of the cortical dorsal visual stream and visuospatial working memory (vsWM) network in individuals with schizophrenia (SZ) likely reflects alterations in both excitatory and inhibitory neurotransmission within nodes responsible for information transfer across the network, including primary visual (V1), visual association (V2), posterior parietal (PPC), and dorsolateral prefrontal (DLPFC) cortices. However, the expression patterns of ionotropic glutamatergic and GABAergic receptor subunits across these regions, and alterations of these patterns in SZ, have not been investigated. We quantified transcript levels of key subunits for excitatory N-methyl-D-aspartate receptors (NMDARs), excitatory alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), and inhibitory GABAA receptors (GABAARs) in postmortem total gray matter from V1, V2, PPC, and DLPFC of unaffected comparison (UC) and matched SZ subjects. In UC subjects, levels of most AMPAR and NMDAR mRNAs exhibited opposite rostral-to-caudal gradients, with AMPAR GRIA1 and GRIA2 mRNA levels highest in DLPFC and NMDAR GRIN1 and GRIN2A mRNA levels highest in V1. GABRA5 and GABRA1 mRNA levels were highest in DLPFC and V1, respectively. In SZ, most transcript levels were lower relative to UC subjects, with these differences largest in V1, intermediate in V2 and PPC, and smallest in DLPFC. In UC subjects, these distinct patterns of receptor transcript levels across the cortical vsWM network suggest that the balance between excitation and inhibition is achieved in a region-specific manner. In SZ subjects, the large deficits in excitatory and inhibitory receptor transcript levels in caudal sensory regions suggest that abnormalities early in the vsWM pathway might contribute to altered information processing in rostral higher-order regions.
Collapse
Affiliation(s)
- Kirsten E Schoonover
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Behavioral Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samuel J Dienel
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, USA
| | - H Holly Bazmi
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F Enwright
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Marín O. Parvalbumin interneuron deficits in schizophrenia. Eur Neuropsychopharmacol 2024; 82:44-52. [PMID: 38490084 PMCID: PMC11413553 DOI: 10.1016/j.euroneuro.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/17/2024]
Abstract
Parvalbumin-expressing (PV+) interneurons represent one of the most abundant subclasses of cortical interneurons. Owing to their specific electrophysiological and synaptic properties, PV+ interneurons are essential for gating and pacing the activity of excitatory neurons. In particular, PV+ interneurons are critically involved in generating and maintaining cortical rhythms in the gamma frequency, which are essential for complex cognitive functions. Deficits in PV+ interneurons have been frequently reported in postmortem studies of schizophrenia patients, and alterations in gamma oscillations are a prominent electrophysiological feature of the disease. Here, I summarise the main features of PV+ interneurons and review clinical and preclinical studies linking the developmental dysfunction of cortical PV+ interneurons with the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
16
|
Dummer PD, Lee DI, Hossain S, Wang R, Evard A, Newman G, Ho C, Schneider-Mizell CM, Menon V, Au E. Multidimensional analysis of cortical interneuron synaptic features reveals underlying synaptic heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586340. [PMID: 38659827 PMCID: PMC11042224 DOI: 10.1101/2024.03.22.586340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cortical interneurons represent a diverse set of neuronal subtypes characterized in part by their striking degree of synaptic specificity. However, little is known about the extent of synaptic diversity because of the lack of unbiased methods to extract synaptic features among interneuron subtypes. Here, we develop an approach to aggregate image features from fluorescent confocal images of interneuron synapses and their post-synaptic targets, in order to characterize the heterogeneity of synapses at fine scale. We started by training a model that recognizes pre- and post-synaptic compartments and then determines the target of each genetically-identified interneuron synapse in vitro and in vivo. Our model extracts hundreds of spatial and intensity features from each analyzed synapse, constructing a multidimensional data set, consisting of millions of synapses, which allowed us to perform an unsupervised analysis on this dataset, uncovering novel synaptic subgroups. The subgroups were spatially distributed in a highly structured manner that revealed the local underlying topology of the postsynaptic environment. Dendrite-targeting subgroups were clustered onto subdomains of the dendrite along the proximal to distal axis. Soma-targeting subgroups were enriched onto different postsynaptic cell types. We also find that the two main subclasses of interneurons, basket cells and somatostatin interneurons, utilize distinct strategies to enact inhibitory coverage. Thus, our analysis of multidimensional synaptic features establishes a conceptual framework for studying interneuron synaptic diversity.
Collapse
Affiliation(s)
- Patrick D. Dummer
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Dylan I. Lee
- Department of Neurology, Columbia Irving Medical Center, New York NY, 10032
| | - Sakib Hossain
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Runsheng Wang
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Andre Evard
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Gabriel Newman
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Claire Ho
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | | | - Vilas Menon
- Department of Neurology, Columbia Irving Medical Center, New York NY, 10032
| | - Edmund Au
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
- Columbia Translation Neuroscience Initiative Fellow, Columbia Irving Medical Center, New York NY, 10032
| |
Collapse
|
17
|
Negrón-Oyarzo I, Dib T, Chacana-Véliz L, López-Quilodrán N, Urrutia-Piñones J. Large-scale coupling of prefrontal activity patterns as a mechanism for cognitive control in health and disease: evidence from rodent models. Front Neural Circuits 2024; 18:1286111. [PMID: 38638163 PMCID: PMC11024307 DOI: 10.3389/fncir.2024.1286111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Cognitive control of behavior is crucial for well-being, as allows subject to adapt to changing environments in a goal-directed way. Changes in cognitive control of behavior is observed during cognitive decline in elderly and in pathological mental conditions. Therefore, the recovery of cognitive control may provide a reliable preventive and therapeutic strategy. However, its neural basis is not completely understood. Cognitive control is supported by the prefrontal cortex, structure that integrates relevant information for the appropriate organization of behavior. At neurophysiological level, it is suggested that cognitive control is supported by local and large-scale synchronization of oscillatory activity patterns and neural spiking activity between the prefrontal cortex and distributed neural networks. In this review, we focus mainly on rodent models approaching the neuronal origin of these prefrontal patterns, and the cognitive and behavioral relevance of its coordination with distributed brain systems. We also examine the relationship between cognitive control and neural activity patterns in the prefrontal cortex, and its role in normal cognitive decline and pathological mental conditions. Finally, based on these body of evidence, we propose a common mechanism that may underlie the impaired cognitive control of behavior.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Tatiana Dib
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Chacana-Véliz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Nélida López-Quilodrán
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jocelyn Urrutia-Piñones
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
18
|
Thompson SM. Modulators of GABA A receptor-mediated inhibition in the treatment of neuropsychiatric disorders: past, present, and future. Neuropsychopharmacology 2024; 49:83-95. [PMID: 37709943 PMCID: PMC10700661 DOI: 10.1038/s41386-023-01728-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
The predominant inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), acts at ionotropic GABAA receptors to counterbalance excitation and regulate neuronal firing. GABAA receptors are heteropentameric channels comprised from subunits derived from 19 different genes. GABAA receptors have one of the richest and well-developed pharmacologies of any therapeutic drug target, including agonists, antagonists, and positive and negative allosteric modulators (PAMs, NAMs). Currently used PAMs include benzodiazepine sedatives and anxiolytics, barbiturates, endogenous and synthetic neurosteroids, and general anesthetics. In this article, I will review evidence that these drugs act at several distinct binding sites and how they can be used to alter the balance between excitation and inhibition. I will also summarize existing literature regarding (1) evidence that changes in GABAergic inhibition play a causative role in major depression, anxiety, postpartum depression, premenstrual dysphoric disorder, and schizophrenia and (2) whether and how GABAergic drugs exert beneficial effects in these conditions, focusing on human studies where possible. Where these classical therapeutics have failed to exert benefits, I will describe recent advances in clinical and preclinical drug development. I will also highlight opportunities to advance a generation of GABAergic therapeutics, such as development of subunit-selective PAMs and NAMs, that are engendering hope for novel tools to treat these devastating conditions.
Collapse
Affiliation(s)
- Scott M Thompson
- Center for Novel Therapeutics, Department of Psychiatry, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
19
|
Clarin JD, Reddy N, Alexandropoulos C, Gao WJ. The role of cell adhesion molecule IgSF9b at the inhibitory synapse and psychiatric disease. Neurosci Biobehav Rev 2024; 156:105476. [PMID: 38029609 PMCID: PMC10842117 DOI: 10.1016/j.neubiorev.2023.105476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/15/2023] [Accepted: 11/18/2023] [Indexed: 12/01/2023]
Abstract
Understanding perturbations in synaptic function between health and disease states is crucial to the treatment of neuropsychiatric illness. While genome-wide association studies have identified several genetic loci implicated in synaptic dysfunction in disorders such as autism and schizophrenia, many have not been rigorously characterized. Here, we highlight immunoglobulin superfamily member 9b (IgSF9b), a cell adhesion molecule thought to localize exclusively to inhibitory synapses in the brain. While both pre-clinical and clinical studies suggest its association with psychiatric diseases, our understanding of IgSF9b in synaptic maintenance, neural circuits, and behavioral phenotypes remains rudimentary. Moreover, these functions wield undiscovered influences on neurodevelopment. This review evaluates current literature and publicly available gene expression databases to explore the implications of IgSF9b dysfunction in rodents and humans. Through a focused analysis of one high-risk gene locus, we identify areas requiring further investigation and unearth clues related to broader mechanisms contributing to the synaptic etiology of psychiatric disorders.
Collapse
Affiliation(s)
- Jacob D Clarin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Natasha Reddy
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Cassandra Alexandropoulos
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States.
| |
Collapse
|
20
|
Gillespie B, Panthi S, Sundram S, Hill RA. The impact of maternal immune activation on GABAergic interneuron development: A systematic review of rodent studies and their translational implications. Neurosci Biobehav Rev 2024; 156:105488. [PMID: 38042358 DOI: 10.1016/j.neubiorev.2023.105488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Mothers exposed to infections during pregnancy disproportionally birth children who develop autism and schizophrenia, disorders associated with altered GABAergic function. The maternal immune activation (MIA) model recapitulates this risk factor, with many studies also reporting disruptions to GABAergic interneuron expression, protein, cellular density and function. However, it is unclear if there are species, sex, age, region, or GABAergic subtype specific vulnerabilities to MIA. Furthermore, to fully comprehend the impact of MIA on the GABAergic system a synthesised account of molecular, cellular, electrophysiological and behavioural findings was required. To this end we conducted a systematic review of GABAergic interneuron changes in the MIA model, focusing on the prefrontal cortex and hippocampus. We reviewed 102 articles that revealed robust changes in a number of GABAergic markers that present as gestationally-specific, region-specific and sometimes sex-specific. Disruptions to GABAergic markers coincided with distinct behavioural phenotypes, including memory, sensorimotor gating, anxiety, and sociability. Findings suggest the MIA model is a valid tool for testing novel therapeutics designed to recover GABAergic function and associated behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Sandesh Panthi
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
21
|
Cameron D, Vinh NN, Prapaiwongs P, Perry EA, Walters JTR, Li M, O’Donovan MC, Bray NJ. Genetic implication of prenatal GABAergic and cholinergic neuron development in susceptibility to schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.14.23299948. [PMID: 38168283 PMCID: PMC10760267 DOI: 10.1101/2023.12.14.23299948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background The ganglionic eminences are fetal-specific structures that give rise to gamma-aminobutyric acid (GABA)- and acetylcholine- releasing neurons of the forebrain. Given evidence for GABAergic and cholinergic disturbances in schizophrenia, as well as an early neurodevelopmental component to the disorder, we tested the potential involvement of developing cells of the ganglionic eminences in mediating genetic risk for the condition. Study Design We combined data from a recent large-scale genome-wide association study of schizophrenia with single cell RNA sequencing data from the human ganglionic eminences to test enrichment of schizophrenia risk variation in genes with high expression specificity for particular developing cell populations within these structures. We additionally performed the single nuclei Assay for Transposase-Accessible Chromatin with Sequencing (snATAC-Seq) to map potential regulatory genomic regions operating in individual cell populations of the human ganglionic eminences, using these to additionally test for enrichment of schizophrenia common genetic variant liability and to functionally annotate non-coding variants associated with the disorder. Study Results Schizophrenia common variant liability was enriched in genes with high expression specificity for developing neuron populations that are predicted to form dopamine D1 and D2 receptor expressing GABAergic medium spiny neurons of the striatum, cortical somatostatin-positive GABAergic interneurons, calretinin-positive GABAergic neurons and cholinergic neurons. Consistent with these findings, schizophrenia genetic risk was also concentrated in predicted regulatory genomic sequence mapped in developing neuronal populations of the ganglionic eminences. Conclusions Our study provides evidence for a role of prenatal GABAergic and cholinergic neuron development in later susceptibility to schizophrenia.
Collapse
Affiliation(s)
- Darren Cameron
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Ngoc-Nga Vinh
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Parinda Prapaiwongs
- Neuroscience & Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
| | - Elizabeth A. Perry
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - James T. R. Walters
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Meng Li
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- Neuroscience & Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
| | - Michael C. O’Donovan
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Nicholas J. Bray
- Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- Neuroscience & Mental Health Innovation Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
22
|
Dienel SJ, Dowling KF, Barile Z, Bazmi HH, Liu A, Vespoli JC, Fish KN, Lewis DA. Diagnostic Specificity and Association With Cognition of Molecular Alterations in Prefrontal Somatostatin Neurons in Schizophrenia. JAMA Psychiatry 2023; 80:1235-1245. [PMID: 37647039 PMCID: PMC10469307 DOI: 10.1001/jamapsychiatry.2023.2972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/14/2023] [Indexed: 09/01/2023]
Abstract
Importance Individuals with schizophrenia (SZ) exhibit pronounced deficits in somatostatin (SST) messenger RNA (mRNA) levels in the dorsolateral prefrontal cortex (DLPFC). Molecularly distinct subtypes of SST neurons, located in the superficial and deep zones of the DLPFC, are thought to contribute to different functional processes of this region; understanding the specificity of SST alterations in SZ across these zones could inform the functional consequences of those alterations, including cognitive impairments characteristic of SZ. Objective To quantify mRNA levels of SST and related neuropeptides in the DLPFC in individuals with SZ, bipolar disorder (BPD), or major depressive disorder (MDD) and unaffected comparison individuals. Design, Setting, and Participants This case-control study, conducted from January 20, 2020, to March 30, 2022, used postmortem brain tissue specimens previously obtained from individuals with SZ, MDD, or BPD and unaffected individuals from a community population through 2 medical examiners' offices. Demographic, clinical, and educational information was ascertained through psychological autopsies. Exposures Diagnosis of SZ, BPD, or MDD. Main Outcome and Measures The main outcome was levels of SST and related neuropeptide mRNA in 2 DLPFC zones, examined using laser microdissection and quantitative polymerase chain reaction or fluorescent in situ hybridization (FISH). Findings were compared using educational attainment as a proxy measure of premorbid cognition. Results A total of 200 postmortem brain specimens were studied, including 65 from unaffected comparison individuals (42 [65%] male; mean [SD] age, 49.2 [14.1] years); 54 from individuals with SZ (37 [69%] male; mean [SD] age, 47.5 [13.3] years); 42 from individuals with MDD (24 [57%] male; mean [SD] age, 45.6 [12.1] years); and 39 from individuals with BPD (23 [59%] male; mean (SD) age, 46.2 [12.5] years). Compared with unaffected individuals, levels of SST mRNA were lower in both superficial (Cohen d, 0.68; 95% CI, 0.23-1.13; P = .004) and deep (Cohen d, 0.60; 95% CI, 0.16-1.04; P = .02) DLPFC zones in individuals with SZ; findings were confirmed using FISH. Levels of SST were lower only in the superficial zone in the group with MDD (Cohen d, 0.58; 95% CI, 0.14-1.02; P = .12), but the difference was not significant; SST levels were not lower in either zone in the BPD group. Levels of neuropeptide Y and tachykinin 1 showed similar patterns. Neuropeptide alterations in the superficial, but not deep, zone were associated with lower educational attainment only in the group with SZ (superficial: adjusted odds ratio, 1.71 [95% CI, 1.11-2.69]; P = .02; deep: adjusted odds ratio, 1.08 [95% CI, 0.64-1.84]; P = .77). Conclusions and Relevance The findings revealed diagnosis-specific patterns of molecular alterations in SST neurons in the DLPFC, suggesting that distinct disease processes are reflected in the differential vulnerability of SST neurons in individuals with SZ, MDD, and BPD. In SZ, alterations specifically in the superficial zone may be associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Samuel J. Dienel
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kevin F. Dowling
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zackery Barile
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - H. Holly Bazmi
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amy Liu
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Julia C. Vespoli
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kenneth N. Fish
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A. Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
23
|
Usui K, Kirihara K, Araki T, Tada M, Koshiyama D, Fujioka M, Nishimura R, Ando S, Koike S, Sugiyama H, Shirakawa T, Toriyama R, Masaoka M, Fujikawa S, Endo K, Yamasaki S, Nishida A, Kasai K. Longitudinal change in mismatch negativity (MMN) but not in gamma-band auditory steady-state response (ASSR) is associated with psychological difficulties in adolescence. Cereb Cortex 2023; 33:11070-11079. [PMID: 37815245 PMCID: PMC10631957 DOI: 10.1093/cercor/bhad346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/26/2023] [Accepted: 08/21/2023] [Indexed: 10/11/2023] Open
Abstract
Adolescence is a critical period for psychological difficulties. Auditory mismatch negativity (MMN) and gamma-band auditory steady-state response (ASSR) are representative electrophysiological indices that mature during adolescence. However, the longitudinal association between MMN/ASSR and psychological difficulties among adolescents remains unclear. We measured MMN amplitude for duration and frequency changes and ASSR twice in a subsample (n = 67, mean age 13.4 and 16.1 years, respectively) from a large-scale population-based cohort. No significant longitudinal changes were observed in any of the electroencephalography indices. Changes in SDQ-TD were significantly associated with changes in duration MMN, but not frequency MMN and ASSR. Furthermore, the subgroup with higher SDQ-TD at follow-up showed a significant duration MMN decrease over time, whereas the subgroup with lower SDQ-TD did not. The results of our population neuroscience study suggest that insufficient changes in electroencephalography indices may have been because of the short follow-up period or non-monotonic change during adolescence, and indicated that the longitudinal association with psychological difficulties was specific to the duration MMN. These findings provide new insights that electrophysiological change may underlie the development of psychosocial difficulties emerging in adolescence.
Collapse
Affiliation(s)
- Kaori Usui
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Department of Community Mental Health & Law, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, 187-8551, Japan
| | - Kenji Kirihara
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Disability Services Office, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Tsuyoshi Araki
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Department of Neuropsychiatry, Teikyo University Hospital, Mizonokuchi, Tokyo, 213-8507, Japan
| | - Mariko Tada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Office for Mental Health Support, Center for Research on Counseling and Support Services, The University of Tokyo, Tokyo, 113-8655, Japan
- The International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
| | - Daisuke Koshiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Mao Fujioka
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Ryoichi Nishimura
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Shuntaro Ando
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shinsuke Koike
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- The International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM), Tokyo, 113-8655, Japan
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, 153-8902, Japan
| | - Hiroshi Sugiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Toru Shirakawa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Department of Electrical Engineering and Computer Science, Faculty of Systems Design, Tokyo Metropolitan University, Tokyo, 192-0397 Japan
| | - Rie Toriyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Mio Masaoka
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Shinya Fujikawa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Kaori Endo
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Syudo Yamasaki
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsushi Nishida
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- The International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
- University of Tokyo Institute for Diversity & Adaptation of Human Mind (UTIDAHM), Tokyo, 113-8655, Japan
| |
Collapse
|
24
|
Miyahara K, Hino M, Shishido R, Izumi R, Nagaoka A, Hayashi H, Kakita A, Yabe H, Tomita H, Kunii Y. Ethnicity-dependent effect of rs1799971 polymorphism on OPRM1 expression in the postmortem brain and responsiveness to antipsychotics. J Psychiatr Res 2023; 166:10-16. [PMID: 37659266 DOI: 10.1016/j.jpsychires.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/25/2023] [Indexed: 09/04/2023]
Abstract
Schizophrenia is associated with aberration of inhibitory neurons. Although the mu-opioid receptor (MOR) is an essential modulator of inhibitory neurons, the effect of rs1799971 polymorphism in the MOR gene on risk of schizophrenia is controversial. Moreover, the disturbance of opioids systems in patients with schizophrenia has not been fully examined. We firstly conducted preliminary meta-analyses integrating Asian and European populations separately over 12,000 subjects to assess the effect of rs1799971 on risk of schizophrenia. Based on the above result, we also investigated the effect on the expression levels of MOR mRNA in the prefrontal cortex (PFC) and caudate nucleus of 41 postmortem brains. In addition, we determined whether these levels were related to antemortem schizophrenia symptoms and pharmacotherapeutic effects. The rs1799971 G-allele reduced the risk of schizophrenia in Asian populations (OR: 0.56, 95%CI: 0.32-0.98, p = 0.042) but increased it in European populations (OR: 1.66, 95%CI: 1.08-2.56, p = 0.022). It decreased MOR mRNA levels in PFC in the Japanese population (p = 0.031). Increased MOR mRNA level in PFC correlated with higher total score of antemortem schizophrenia symptoms (p = 0.017). Furthermore, the pharmacotherapeutic effect of first-generation antipsychotics was higher for genotype AA than AG/GG of rs1799971 (p = 0.036). The rs1799971 affects risk of schizophrenia and MOR mRNA expression and the effect varies according to ethnicity. Overexpression of MOR might induce severe schizophrenia symptoms. Therefore, MOR modulation may be the key clue for treating antipsychotics-resistant schizophrenia, and genotyping rs1799971 may provide a better pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- Kazusa Miyahara
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Mizuki Hino
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan; Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Risa Shishido
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Ryuta Izumi
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Atsuko Nagaoka
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hideki Hayashi
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Tohoku University Hospital, Miyagi, Japan; Department of Psychiatry, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Yasuto Kunii
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan; Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| |
Collapse
|
25
|
Ollà I, Pardiñas AF, Parras A, Hernández IH, Santos-Galindo M, Picó S, Callado LF, Elorza A, Rodríguez-López C, Fernández-Miranda G, Belloc E, Walters JTR, O'Donovan MC, Méndez R, Toma C, Meana JJ, Owen MJ, Lucas JJ. Pathogenic Mis-splicing of CPEB4 in Schizophrenia. Biol Psychiatry 2023; 94:341-351. [PMID: 36958377 DOI: 10.1016/j.biopsych.2023.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Schizophrenia (SCZ) is caused by an interplay of polygenic risk and environmental factors, which may alter regulators of gene expression leading to pathogenic misexpression of SCZ risk genes. The CPEB family of RNA-binding proteins (CPEB1-4) regulates translation of target RNAs (approximately 40% of overall genes). We previously identified CPEB4 as a key dysregulated translational regulator in autism spectrum disorder (ASD) because its neuronal-specific microexon (exon 4) is mis-spliced in ASD brains, causing underexpression of numerous ASD risk genes. The genetic factors and pathogenic mechanisms shared between SCZ and ASD led us to hypothesize CPEB4 mis-splicing in SCZ leading to underexpression of multiple SCZ-related genes. METHODS We performed MAGMA-enrichment analysis on Psychiatric Genomics Consortium genome-wide association study data and analyzed RNA sequencing data from the PsychENCODE Consortium. Reverse transcriptase polymerase chain reaction and Western blot were performed on postmortem brain tissue, and the presence/absence of antipsychotics was assessed through toxicological analysis. Finally, mice with mild overexpression of exon 4-lacking CPEB4 (CPEB4Δ4) were generated and analyzed biochemically and behaviorally. RESULTS First, we found enrichment of SCZ-associated genes for CPEB4-binder transcripts. We also found decreased usage of CPEB4 microexon in SCZ probands, which was correlated with decreased protein levels of CPEB4-target SCZ-associated genes only in antipsychotic-free individuals. Interestingly, differentially expressed genes fit those reported for SCZ, specifically in the SCZ probands with decreased CPEB4-microexon inclusion. Finally, we demonstrated that mice with mild overexpression of CPEB4Δ4 showed decreased protein levels of CPEB4-target SCZ genes and SCZ-linked behaviors. CONCLUSIONS We identified aberrant CPEB4 splicing and downstream misexpression of SCZ risk genes as a novel etiological mechanism in SCZ.
Collapse
Affiliation(s)
- Ivana Ollà
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio F Pardiñas
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Alberto Parras
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Ivó H Hernández
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - María Santos-Galindo
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Picó
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Biocruces Bizkaia Health Research Institute and Networking Research Center on Mental Health (Centro de investigación Biomédica en Red | Salud Mental), Leioa, Bizkaia, Spain
| | - Ainara Elorza
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Claudia Rodríguez-López
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo Fernández-Miranda
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Eulàlia Belloc
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - James T R Walters
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Raúl Méndez
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain; Institució Catalana de RIcerca i Estudis Avançats, Barcelona, Spain
| | - Claudio Toma
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Biocruces Bizkaia Health Research Institute and Networking Research Center on Mental Health (Centro de investigación Biomédica en Red | Salud Mental), Leioa, Bizkaia, Spain
| | - Michael J Owen
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - José J Lucas
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
26
|
Dowling KF, Dienel SJ, Barile Z, Bazmi HH, Lewis DA. Localization and Diagnostic Specificity of Glutamic Acid Decarboxylase Transcript Alterations in the Dorsolateral Prefrontal Cortex in Schizophrenia. Biol Psychiatry 2023; 94:322-331. [PMID: 37061080 PMCID: PMC10524522 DOI: 10.1016/j.biopsych.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Working memory (WM) deficits in schizophrenia are thought to reflect altered inhibition in the dorsolateral prefrontal cortex (DLPFC). This interpretation is supported by findings of lower transcript levels of the 2 enzymes, GAD67 and GAD65, which mediate basal and activity-dependent GABA (gamma-aminobutyric acid) synthesis, respectively. However, the relative magnitude, location within the depth of the DLPFC, and specificity to the disease process of schizophrenia of alterations in GAD67 and/or GAD65 remain unclear. METHODS Levels of GAD67 and GAD65 messenger RNAs (mRNAs) in superficial (layers 2/superficial 3) and deep (deep layer 6/white matter) zones of the DLPFC were quantified by quantitative polymerase chain reaction in subjects with schizophrenia (n = 41), major depression (n = 42), or bipolar disorder (n = 39) and unaffected comparison (n = 43) subjects. RESULTS Relative to the unaffected comparison group, GAD67 and GAD65 mRNA levels in the schizophrenia group were lower (p = .039, effect size = -0.69 and p = .027, effect size = -0.72, respectively) in the superficial zone but were unaltered in the deep zone. In the major depression group, only GAD67 mRNA levels were lower and only in the superficial zone (p = .089, effect size = 0.70). No differences were detected in the bipolar disorder group. Neither GAD67 nor GAD65 mRNA alterations were explained by psychosis, mood disturbance, or common comorbid factors. CONCLUSIONS Alterations in markers of GABA synthesis demonstrated transcript, DLPFC zone, and diagnostic specificity. Given the dependence of WM on GABA neurotransmission in the superficial DLPFC, our findings suggest that limitations to GABA synthesis in this location contribute to WM impairments in schizophrenia, especially during demanding WM tasks, when GABA synthesis requires the activity of both GAD67 and GAD65.
Collapse
Affiliation(s)
- Kevin F Dowling
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Samuel J Dienel
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Zackery Barile
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - H Holly Bazmi
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
27
|
Mahadevan AS, Cornblath EJ, Lydon-Staley DM, Zhou D, Parkes L, Larsen B, Adebimpe A, Kahn AE, Gur RC, Gur RE, Satterthwaite TD, Wolf DH, Bassett DS. Alprazolam modulates persistence energy during emotion processing in first-degree relatives of individuals with schizophrenia: a network control study. Mol Psychiatry 2023; 28:3314-3323. [PMID: 37353585 PMCID: PMC10618098 DOI: 10.1038/s41380-023-02121-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/28/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023]
Abstract
Schizophrenia is marked by deficits in facial affect processing associated with abnormalities in GABAergic circuitry, deficits also found in first-degree relatives. Facial affect processing involves a distributed network of brain regions including limbic regions like amygdala and visual processing areas like fusiform cortex. Pharmacological modulation of GABAergic circuitry using benzodiazepines like alprazolam can be useful for studying this facial affect processing network and associated GABAergic abnormalities in schizophrenia. Here, we use pharmacological modulation and computational modeling to study the contribution of GABAergic abnormalities toward emotion processing deficits in schizophrenia. Specifically, we apply principles from network control theory to model persistence energy - the control energy required to maintain brain activation states - during emotion identification and recall tasks, with and without administration of alprazolam, in a sample of first-degree relatives and healthy controls. Here, persistence energy quantifies the magnitude of theoretical external inputs during the task. We find that alprazolam increases persistence energy in relatives but not in controls during threatening face processing, suggesting a compensatory mechanism given the relative absence of behavioral abnormalities in this sample of unaffected relatives. Further, we demonstrate that regions in the fusiform and occipital cortices are important for facilitating state transitions during facial affect processing. Finally, we uncover spatial relationships (i) between regional variation in differential control energy (alprazolam versus placebo) and (ii) both serotonin and dopamine neurotransmitter systems, indicating that alprazolam may exert its effects by altering neuromodulatory systems. Together, these findings provide a new perspective on the distributed emotion processing network and the effect of GABAergic modulation on this network, in addition to identifying an association between schizophrenia risk and abnormal GABAergic effects on persistence energy during threat processing.
Collapse
Affiliation(s)
- Arun S Mahadevan
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eli J Cornblath
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - David M Lydon-Staley
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Annenberg School for Communication, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dale Zhou
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Linden Parkes
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bart Larsen
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Azeez Adebimpe
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ari E Kahn
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Ruben C Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Raquel E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
| | - Theodore D Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel H Wolf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dani S Bassett
- Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Electrical & Systems Engineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Physics & Astronomy, College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Santa Fe Institute, 1399 Hyde Park Rd, Santa Fe, NM, 87501, USA.
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
28
|
Fish KN, Rocco BR, Wilson JD, Lewis DA. Laminar-Specific Alterations in Calbindin-Positive Boutons in the Prefrontal Cortex of Subjects With Schizophrenia. Biol Psychiatry 2023; 94:142-152. [PMID: 36868891 PMCID: PMC10247897 DOI: 10.1016/j.biopsych.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cognitive deficits in schizophrenia are associated with altered GABA (gamma-aminobutyric acid) neurotransmission in the prefrontal cortex (PFC). GABA neurotransmission requires GABA synthesis by 2 isoforms of glutamic acid decarboxylase (GAD65 and GAD67) and packaging by the vesicular GABA transporter (vGAT). Current postmortem findings suggest that GAD67 messenger RNA is lower in a subset of the calbindin-expressing (CB+) class of GABA neurons in schizophrenia. Hence, we assessed if CB+ GABA neuron boutons are affected in schizophrenia. METHODS For 20 matched pairs of subjects with schizophrenia and unaffected comparison subjects, PFC tissue sections were immunolabeled for vGAT, CB, GAD67, and GAD65. The density of CB+ GABA boutons and levels of the 4 proteins per bouton were quantified. RESULTS Some CB+ GABA boutons contained both GAD65 and GAD67 (GAD65+/GAD67+), whereas others contained only GAD65 (GAD65+) or GAD67 (GAD67+). In schizophrenia, vGAT+/CB+/GAD65+/GAD67+ bouton density was not altered, vGAT+/CB+/GAD65+ bouton density was 86% higher in layers 2/superficial 3 (L2/3s), and vGAT+/CB+/GAD67+ bouton density was 36% lower in L5-6. Bouton GAD levels were differentially altered across bouton types and layers. In schizophrenia, the sum of GAD65 and GAD67 levels in vGAT+/CB+/GAD65+/GAD67+ boutons was 36% lower in L6, GAD65 levels were 51% higher in vGAT+/CB+/GAD65+ boutons in L2, and GAD67 levels in vGAT+/CB+/GAD67+ boutons were 30% to 46% lower in L2/3s-6. CONCLUSIONS These findings indicate that schizophrenia-associated alterations in the strength of inhibition from CB+ GABA neurons in the PFC differ across cortical layers and bouton classes, suggesting complex contributions to PFC dysfunction and cognitive impairments in schizophrenia.
Collapse
Affiliation(s)
- Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Brad R Rocco
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James D Wilson
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
29
|
Vid Prkačin M, Banovac I, Petanjek Z, Hladnik A. Cortical interneurons in schizophrenia - cause or effect? Croat Med J 2023; 64:110-122. [PMID: 37131313 PMCID: PMC10183954 DOI: 10.3325/cmj.2023.64.110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/15/2023] [Indexed: 12/09/2024] Open
Abstract
GABAergic cortical interneurons are important components of cortical microcircuits. Their alterations are associated with a number of neurological and psychiatric disorders, and are thought to be especially important in the pathogenesis of schizophrenia. Here, we reviewed neuroanatomical and histological studies that analyzed different populations of cortical interneurons in postmortem human tissue from patients with schizophrenia and adequately matched controls. The data strongly suggests that in schizophrenia only selective interneuron populations are affected, with alterations of somatostatin and parvalbumin neurons being the most convincing. The most prominent changes are found in the prefrontal cortex, which is consistent with the impairment of higher cognitive functions characteristic of schizophrenia. In contrast, calretinin neurons, the most numerous interneuron population in primates, seem to be largely unaffected. The selective alterations of cortical interneurons are in line with the neurodevelopmental model and the multiple-hit hypothesis of schizophrenia. Nevertheless, a large number of data on interneurons in schizophrenia is still inconclusive, with different studies yielding opposing findings. Furthermore, no studies found a clear link between interneuron alterations and clinical outcomes. Future research should focus on the causes of changes in the cortical microcircuitry in order to identify potential therapeutic targets.
Collapse
Affiliation(s)
| | - Ivan Banovac
- Ivan Banovac, Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, 10 000 Zagreb, Croatia,
| | | | | |
Collapse
|
30
|
Adraoui FW, Douw L, Martens GJM, Maas DA. Connecting Neurobiological Features with Interregional Dysconnectivity in Social-Cognitive Impairments of Schizophrenia. Int J Mol Sci 2023; 24:ijms24097680. [PMID: 37175387 PMCID: PMC10177877 DOI: 10.3390/ijms24097680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Schizophrenia (SZ) is a devastating psychiatric disorder affecting about 1% of the world's population. Social-cognitive impairments in SZ prevent positive social interactions and lead to progressive social withdrawal. The neurobiological underpinnings of social-cognitive symptoms remain poorly understood, which hinders the development of novel treatments. At the whole-brain level, an abnormal activation of social brain regions and interregional dysconnectivity within social-cognitive brain networks have been identified as major contributors to these symptoms. At the cellular and subcellular levels, an interplay between oxidative stress, neuroinflammation and N-methyl-D-aspartate receptor hypofunction is thought to underly SZ pathology. However, it is not clear how these molecular processes are linked with interregional dysconnectivity in the genesis of social-cognitive symptoms. Here, we aim to bridge the gap between macroscale (connectivity analyses) and microscale (molecular and cellular mechanistic) knowledge by proposing impaired myelination and the disinhibition of local microcircuits as possible causative biological pathways leading to dysconnectivity and abnormal activity of the social brain. Furthermore, we recommend electroencephalography as a promising translational technique that can foster pre-clinical drug development and discuss attractive drug targets for the treatment of social-cognitive symptoms in SZ.
Collapse
Affiliation(s)
- Florian W Adraoui
- Biotrial, Preclinical Pharmacology Department, 7-9 rue Jean-Louis Bertrand, 35000 Rennes, France
| | - Linda Douw
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| | - Gerard J M Martens
- Donders Centre for Neuroscience (DCN), Department of Molecular Animal Physiology, Faculty of Science, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 GA Nijmegen, The Netherlands
- NeuroDrug Research Ltd., 6525 ED Nijmegen, The Netherlands
| | - Dorien A Maas
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
31
|
Zhang HC, Du Y, Chen L, Yuan ZQ, Cheng Y. MicroRNA schizophrenia: Etiology, biomarkers and therapeutic targets. Neurosci Biobehav Rev 2023; 146:105064. [PMID: 36707012 DOI: 10.1016/j.neubiorev.2023.105064] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/11/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
The three sets of symptoms associated with schizophrenia-positive, negative, and cognitive-are burdensome and have serious effects on public health, which affects up to 1% of the population. It is now commonly believed that in addition to the traditional dopaminergic mesolimbic pathway, the etiology of schizophrenia also includes neuronal networks, such as glutamate, GABA, serotonin, BDNF, oxidative stress, inflammation and the immune system. Small noncoding RNA molecules called microRNAs (miRNAs) have come to light as possible participants in the pathophysiology of schizophrenia in recent years by having an impact on these systems. These small RNAs regulate the stability and translation of hundreds of target transcripts, which has an impact on the entire gene network. There may be improved approaches to treat and diagnose schizophrenia if it is understood how these changes in miRNAs alter the critical related signaling pathways that drive the development and progression of the illness.
Collapse
Affiliation(s)
- Heng-Chang Zhang
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Zeng-Qiang Yuan
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China; Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China; Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China; Institute of National Security, Minzu University of China, Beijing, China.
| |
Collapse
|
32
|
Raymann S, Schalbetter SM, Schaer R, Bernhardt AC, Mueller FS, Meyer U, Weber-Stadlbauer U. Late prenatal immune activation in mice induces transgenerational effects via the maternal and paternal lineages. Cereb Cortex 2023; 33:2273-2286. [PMID: 36857721 DOI: 10.1093/cercor/bhac207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
Prenatal exposure to infectious or noninfectious immune activation is an environmental risk factor for neurodevelopmental disorders and mental illnesses. Recent research using animal models suggests that maternal immune activation (MIA) during early to middle stages of pregnancy can induce transgenerational effects on brain and behavior, likely via inducing stable epigenetic modifications across generations. Using a mouse model of viral-like MIA, which is based on gestational treatment with poly(I:C), the present study explored whether transgenerational effects can also emerge when MIA occurs in late pregnancy. Our findings demonstrate that the direct descendants born to poly(I:C)-treated mothers display deficits in temporal order memory, which are similarly present in second- and third-generation offspring. These transgenerational effects were mediated via both the maternal and paternal lineages and were accompanied by transient changes in maternal care. In addition to the cognitive effects, late prenatal immune activation induced generation-spanning effects on the prefrontal expression of gamma-aminobutyric acid (GABA)ergic genes, including parvalbumin and distinct alpha-subunits of the GABAA receptor. Together, our results suggest that MIA in late pregnancy has the potential to affect cognitive functions and prefrontal gene expression patterns in multiple generations, highlighting its role in shaping disease risk across generations.
Collapse
Affiliation(s)
- Stephanie Raymann
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Ron Schaer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Alexandra C Bernhardt
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
33
|
Fujihara K. Beyond the γ-aminobutyric acid hypothesis of schizophrenia. Front Cell Neurosci 2023; 17:1161608. [PMID: 37168420 PMCID: PMC10165250 DOI: 10.3389/fncel.2023.1161608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/04/2023] [Indexed: 05/13/2023] Open
Abstract
Abnormalities in the γ-aminobutyric acid (GABA) system have been reported in the postmortem brains of individuals with schizophrenia. In particular, the reduction of one of the GABA-synthesizing enzymes, the 67-kDa isoform of glutamate decarboxylase (GAD67), has garnered interest among researchers because of its role in the formation of γ-oscillations and its potential involvement in the cognitive dysfunction observed in schizophrenia. Although several animal models have been generated to simulate the alterations observed in postmortem brain studies, they exhibit inconsistent behavioral phenotypes, leading to conflicting views regarding their contributions to the pathogenesis and manifestation of schizophrenia symptoms. For instance, GAD67 knockout rats (also known as Gad1 knockout rats) exhibit marked impairments in spatial working memory, but other model animals do not. In this review, we summarize the phenotypic attributes of these animal models and contemplate the potential for secondary modifications that may arise from the disruption of the GABAergic nervous system.
Collapse
Affiliation(s)
- Kazuyuki Fujihara
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
- *Correspondence: Kazuyuki Fujihara,
| |
Collapse
|
34
|
Dorsolateral Prefrontal Cortex Glutamate/Gamma-Aminobutyric Acid (GABA) Alterations in Clinical High Risk and First-Episode Schizophrenia: A Preliminary 7-T Magnetic Resonance Spectroscopy Imaging Study. Int J Mol Sci 2022; 23:ijms232415846. [PMID: 36555487 PMCID: PMC9781166 DOI: 10.3390/ijms232415846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Converging lines of evidence suggest that an imbalance between excitation and inhibition is present in the dorsolateral prefrontal cortex (DLPFC) of schizophrenia (SCZ). Gamma-aminobutyric-acid (GABA) and, to a lesser extent, glutamate (Glu) abnormalities were reported in the DLPFC of SCZ patients, especially on the right hemisphere, by post-mortem studies. However, in vivo evidence of GABA, Glu, and Glu/GABA DLPFC abnormalities, particularly on the right side and the early stages of illness, is limited. In this preliminary study, we utilized 7-Tesla magnetic resonance spectroscopic imaging (MRSI) to investigate bilateral Glu/Creatine (Cre), GABA/Cre, and Glu/GABA in the DLPFC of sixteen first episode schizophrenia (FES), seventeen clinical high risk (CHR), and twenty-six healthy comparison (HC) subjects. FES and CHR had abnormal GABA/Cre and Glu/GABA in the right DLPFC (rDLPFC) compared with HC participants, while no differences were observed in the left DLPFC (lDLPFC) among the three groups. Furthermore, HC had higher Glu/GABA in rDLPFC compared to lDLPFC (R > L), whereas the opposite relationship (R < L) was observed in the DLPFC Glu/GABA of FES patients. Altogether, these findings indicate that GABA/Cre and Glu/GABA DLPFC alterations are present before illness manifestation and worsen in FES patients, thus representing a putative early pathophysiological biomarker for SCZ and related psychotic disorders.
Collapse
|
35
|
Batiuk MY, Tyler T, Dragicevic K, Mei S, Rydbirk R, Petukhov V, Deviatiiarov R, Sedmak D, Frank E, Feher V, Habek N, Hu Q, Igolkina A, Roszik L, Pfisterer U, Garcia-Gonzalez D, Petanjek Z, Adorjan I, Kharchenko PV, Khodosevich K. Upper cortical layer-driven network impairment in schizophrenia. SCIENCE ADVANCES 2022; 8:eabn8367. [PMID: 36223459 PMCID: PMC9555788 DOI: 10.1126/sciadv.abn8367] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 08/24/2022] [Indexed: 05/31/2023]
Abstract
Schizophrenia is one of the most widespread and complex mental disorders. To characterize the impact of schizophrenia, we performed single-nucleus RNA sequencing (snRNA-seq) of >220,000 neurons from the dorsolateral prefrontal cortex of patients with schizophrenia and matched controls. In addition, >115,000 neurons were analyzed topographically by immunohistochemistry. Compositional analysis of snRNA-seq data revealed a reduction in abundance of GABAergic neurons and a concomitant increase in principal neurons, most pronounced for upper cortical layer subtypes, which was substantiated by histological analysis. Many neuronal subtypes showed extensive transcriptomic changes, the most marked in upper-layer GABAergic neurons, including down-regulation in energy metabolism and up-regulation in neurotransmission. Transcription factor network analysis demonstrated a developmental origin of transcriptomic changes. Last, Visium spatial transcriptomics further corroborated upper-layer neuron vulnerability in schizophrenia. Overall, our results point toward general network impairment within upper cortical layers as a core substrate associated with schizophrenia symptomatology.
Collapse
Affiliation(s)
- Mykhailo Y. Batiuk
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Teadora Tyler
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest H-1085, Hungary
| | - Katarina Dragicevic
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Shenglin Mei
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Rasmus Rydbirk
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Viktor Petukhov
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ruslan Deviatiiarov
- The National Center for Personalized Medicine of Endocrine Diseases, Moscow 115478, Russia
- Kazan Federal University, Kazan 420043, Russia
| | - Dora Sedmak
- Croatian Institute for Brain Research and Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Erzsebet Frank
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest H-1085, Hungary
| | - Virginia Feher
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest H-1085, Hungary
| | - Nikola Habek
- Croatian Institute for Brain Research and Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Qiwen Hu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Anna Igolkina
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- St. Petersburg Polytechnical University, St. Petersburg 195251, Russia
| | - Lilla Roszik
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest H-1085, Hungary
| | - Ulrich Pfisterer
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Diego Garcia-Gonzalez
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Zdravko Petanjek
- Croatian Institute for Brain Research and Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Istvan Adorjan
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest H-1085, Hungary
| | - Peter V. Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
36
|
Jung K, Choi Y, Kwon HB. Cortical control of chandelier cells in neural codes. Front Cell Neurosci 2022; 16:992409. [PMID: 36299494 PMCID: PMC9588934 DOI: 10.3389/fncel.2022.992409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022] Open
Abstract
Various cortical functions arise from the dynamic interplay of excitation and inhibition. GABAergic interneurons that mediate synaptic inhibition display significant diversity in cell morphology, electrophysiology, plasticity rule, and connectivity. These heterogeneous features are thought to underlie their functional diversity. Emerging attention on specific properties of the various interneuron types has emphasized the crucial role of cell-type specific inhibition in cortical neural processing. However, knowledge is still limited on how each interneuron type forms distinct neural circuits and regulates network activity in health and disease. To dissect interneuron heterogeneity at single cell-type precision, we focus on the chandelier cell (ChC), one of the most distinctive GABAergic interneuron types that exclusively innervate the axon initial segments (AIS) of excitatory pyramidal neurons. Here we review the current understanding of the structural and functional properties of ChCs and their implications in behavioral functions, network activity, and psychiatric disorders. These findings provide insights into the distinctive roles of various single-type interneurons in cortical neural coding and the pathophysiology of cortical dysfunction.
Collapse
|
37
|
Liu D, Zinski A, Mishra A, Noh H, Park GH, Qin Y, Olorife O, Park JM, Abani CP, Park JS, Fung J, Sawaqed F, Coyle JT, Stahl E, Bendl J, Fullard JF, Roussos P, Zhang X, Stanton PK, Yin C, Huang W, Kim HY, Won H, Cho JH, Chung S. Impact of schizophrenia GWAS loci converge onto distinct pathways in cortical interneurons vs glutamatergic neurons during development. Mol Psychiatry 2022; 27:4218-4233. [PMID: 35701597 DOI: 10.1038/s41380-022-01654-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 02/07/2023]
Abstract
Remarkable advances have been made in schizophrenia (SCZ) GWAS, but gleaning biological insight from these loci is challenging. Genetic influences on gene expression (e.g., eQTLs) are cell type-specific, but most studies that attempt to clarify GWAS loci's influence on gene expression have employed tissues with mixed cell compositions that can obscure cell-specific effects. Furthermore, enriched SCZ heritability in the fetal brain underscores the need to study the impact of SCZ risk loci in specific developing neurons. MGE-derived cortical interneurons (cINs) are consistently affected in SCZ brains and show enriched SCZ heritability in human fetal brains. We identified SCZ GWAS risk genes that are dysregulated in iPSC-derived homogeneous populations of developing SCZ cINs. These SCZ GWAS loci differential expression (DE) genes converge on the PKC pathway. Their disruption results in PKC hyperactivity in developing cINs, leading to arborization deficits. We show that the fine-mapped GWAS locus in the ATP2A2 gene of the PKC pathway harbors enhancer marks by ATACseq and ChIPseq, and regulates ATP2A2 expression. We also generated developing glutamatergic neurons (GNs), another population with enriched SCZ heritability, and confirmed their functionality after transplantation into the mouse brain. Then, we identified SCZ GWAS risk genes that are dysregulated in developing SCZ GNs. GN-specific SCZ GWAS loci DE genes converge on the ion transporter pathway, distinct from those for cINs. Disruption of the pathway gene CACNA1D resulted in deficits of Ca2+ currents in developing GNs, suggesting compromised neuronal function by GWAS loci pathway deficits during development. This study allows us to identify cell type-specific and developmental stage-specific mechanisms of SCZ risk gene function, and may aid in identifying mechanism-based novel therapeutic targets.
Collapse
Affiliation(s)
- Dongxin Liu
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA.
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.
| | - Amy Zinski
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Akanksha Mishra
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Haneul Noh
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
- Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Gun-Hoo Park
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Yiren Qin
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Oshoname Olorife
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - James M Park
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Chiderah P Abani
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Joy S Park
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Janice Fung
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Farah Sawaqed
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Joseph T Coyle
- Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Eli Stahl
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Jaroslav Bendl
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - John F Fullard
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Panos Roussos
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Mental Illness Research Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, 10468, USA
| | - Xiaolei Zhang
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Patric K Stanton
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Changhong Yin
- Department of Pathology, New York Medical College, Valhalla, NY, 10595, USA
| | - Weihua Huang
- Department of Pathology, New York Medical College, Valhalla, NY, 10595, USA
| | - Hae-Young Kim
- Department of Public Health, New York Medical College, Valhalla, NY, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jun-Hyeong Cho
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Sangmi Chung
- Department of Cell biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA.
- Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
38
|
Gu X, Wang J, Jiang X. miR-124- and let-7-Mediated Reprogram of Human Fibroblasts into SST Interneurons. ACS Chem Neurosci 2022; 13:2755-2765. [PMID: 36074953 DOI: 10.1021/acschemneuro.2c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Many neurological disorders stem from defects in or the loss of specific neurons. Dysfunction of γ-aminobutyric acid (GABA)ergic interneurons may cause a variety of neurological and psychiatric disorders such as epilepsy, autism, Alzheimer's disease, and depression. Unlike other types of neurons, which can be generated relatively easily by direct reprogramming, it is difficult to generate GABAergic neurons by traditional methods. Neuronal transdifferentiation of fibroblasts mediated by nongenomic-integrated adenovirus has many advantages, but the efficiency is low, and there is a lack of studies using human cells as the initial materials. In this study, we explored the feasibility of the conversion of human fibroblasts into neurons through adenovirus-mediated gene expression and found that by introducing two microRNAs, miR-124 and let-7, together with several small chemical compounds, they can effectively generate GABAergic neuron-like cells from human neonatal fibroblasts without reverting to a progenitor cell stage. Most of these cells expressed neuronal markers and were all somatostatin (SST)-positive cells. Therefore, our study provides a relatively safe and efficient method to generate SST interneurons.
Collapse
Affiliation(s)
- Xi Gu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510500, China.,Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350000, China
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350000, China
| | - Xiaodan Jiang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510500, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510500, China
| |
Collapse
|
39
|
da Rocha RH, Fernandes CA, Bignotto TS, Margarido VP, Tencatt LFC, da Graça WJ, Gubiani ÉA. Integrated analysis reveals a new species of Corydoras Lacépède, 1803 (Siluriformes: Callichthyidae) in the lower Iguassu River, Brazil. ORG DIVERS EVOL 2022. [DOI: 10.1007/s13127-021-00534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
40
|
Zachlod D, Bludau S, Cichon S, Palomero-Gallagher N, Amunts K. Combined analysis of cytoarchitectonic, molecular and transcriptomic patterns reveal differences in brain organization across human functional brain systems. Neuroimage 2022; 257:119286. [PMID: 35597401 DOI: 10.1016/j.neuroimage.2022.119286] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/22/2022] [Accepted: 05/05/2022] [Indexed: 01/14/2023] Open
Abstract
Brain areas show specific cellular, molecular, and gene expression patterns that are linked to function, but their precise relationships are largely unknown. To unravel these structure-function relationships, a combined analysis of 53 neurotransmitter receptor genes, receptor densities of six transmitter systems and cytoarchitectonic data of the auditory, somatosensory, visual, motor systems was conducted. Besides covariation of areal gene expression with receptor density, the study reveals specific gene expression patterns in functional systems, which are most prominent for the inhibitory GABAA and excitatory glutamatergic NMDA receptors. Furthermore, gene expression-receptor relationships changed in a systematic manner according to information flow from primary to higher associative areas. The findings shed new light on the relationship of anatomical, functional, and molecular and transcriptomic principles of cortical segregation towards a more comprehensive understanding of human brain organization.
Collapse
Affiliation(s)
- Daniel Zachlod
- Institute of Neurosciences and Medicine (INM-1), Research Centre Jülich, Jülich, Germany.
| | - Sebastian Bludau
- Institute of Neurosciences and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Sven Cichon
- Institute of Neurosciences and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; Department of Biomedicine, University of Basel, Basel, Switzerland; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Nicola Palomero-Gallagher
- Institute of Neurosciences and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; C. & O. Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; Department of Psychiatry, Psychotherapy, and Psychosomatics, Medical Faculty, RWTH Aachen, and JARA - Translational Brain Medicine, Aachen, Germany
| | - Katrin Amunts
- Institute of Neurosciences and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; C. & O. Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
41
|
Park DK, Petshow S, Anisimova M, Barragan EV, Gray JA, Stein IS, Zito K. Reduced d-serine levels drive enhanced non-ionotropic NMDA receptor signaling and destabilization of dendritic spines in a mouse model for studying schizophrenia. Neurobiol Dis 2022; 170:105772. [PMID: 35605760 PMCID: PMC9352378 DOI: 10.1016/j.nbd.2022.105772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 10/31/2022] Open
Abstract
Schizophrenia is a psychiatric disorder that affects over 20 million people globally. Notably, schizophrenia is associated with decreased density of dendritic spines and decreased levels of d-serine, a co-agonist required for opening of the N-methyl-d-aspartate receptor (NMDAR). We hypothesized that lowered d-serine levels associated with schizophrenia would enhance ion flux-independent signaling by the NMDAR, driving destabilization and loss of dendritic spines. We tested our hypothesis using the serine racemase knockout (SRKO) mouse model, which lacks the enzyme for d-serine production. We show that activity-dependent spine growth is impaired in SRKO mice, but can be acutely rescued by exogenous d-serine. Moreover, we find a significant bias of synaptic plasticity toward spine shrinkage in the SRKO mice as compared to wild-type littermates. Notably, we demonstrate that enhanced ion flux-independent signaling through the NMDAR contributes to this bias toward spine destabilization, which is exacerbated by an increase in synaptic NMDARs in hippocampal synapses of SRKO mice. Our results support a model in which lowered d-serine levels associated with schizophrenia enhance ion flux-independent NMDAR signaling and bias toward spine shrinkage and destabilization.
Collapse
|
42
|
Metzner C, Mäki-Marttunen T, Karni G, McMahon-Cole H, Steuber V. The effect of alterations of schizophrenia-associated genes on gamma band oscillations. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:46. [PMID: 35854005 PMCID: PMC9261091 DOI: 10.1038/s41537-022-00255-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
Abnormalities in the synchronized oscillatory activity of neurons in general and, specifically in the gamma band, might play a crucial role in the pathophysiology of schizophrenia. While these changes in oscillatory activity have traditionally been linked to alterations at the synaptic level, we demonstrate here, using computational modeling, that common genetic variants of ion channels can contribute strongly to this effect. Our model of primary auditory cortex highlights multiple schizophrenia-associated genetic variants that reduce gamma power in an auditory steady-state response task. Furthermore, we show that combinations of several of these schizophrenia-associated variants can produce similar effects as the more traditionally considered synaptic changes. Overall, our study provides a mechanistic link between schizophrenia-associated common genetic variants, as identified by genome-wide association studies, and one of the most robust neurophysiological endophenotypes of schizophrenia.
Collapse
Affiliation(s)
- Christoph Metzner
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany.
- Biocomputation Research Group, School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom.
| | | | - Gili Karni
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany
- Minerva Schools at KGI, San Francisco, CA, USA
| | - Hana McMahon-Cole
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany
- Minerva Schools at KGI, San Francisco, CA, USA
| | - Volker Steuber
- Biocomputation Research Group, School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
43
|
Domínguez-Sala E, Valdés-Sánchez L, Canals S, Reiner O, Pombero A, García-López R, Estirado A, Pastor D, Geijo-Barrientos E, Martínez S. Abnormalities in Cortical GABAergic Interneurons of the Primary Motor Cortex Caused by Lis1 (Pafah1b1) Mutation Produce a Non-drastic Functional Phenotype. Front Cell Dev Biol 2022; 10:769853. [PMID: 35309904 PMCID: PMC8924048 DOI: 10.3389/fcell.2022.769853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022] Open
Abstract
LIS1 (PAFAH1B1) plays a major role in the developing cerebral cortex, and haploinsufficient mutations cause human lissencephaly type 1. We have studied morphological and functional properties of the cerebral cortex of mutant mice harboring a deletion in the first exon of the mouse Lis1 (Pafah1b1) gene, which encodes for the LisH domain. The Lis1/sLis1 animals had an overall unaltered cortical structure but showed an abnormal distribution of cortical GABAergic interneurons (those expressing calbindin, calretinin, or parvalbumin), which mainly accumulated in the deep neocortical layers. Interestingly, the study of the oscillatory activity revealed an apparent inability of the cortical circuits to produce correct activity patterns. Moreover, the fast spiking (FS) inhibitory GABAergic interneurons exhibited several abnormalities regarding the size of the action potentials, the threshold for spike firing, the time course of the action potential after-hyperpolarization (AHP), the firing frequency, and the frequency and peak amplitude of spontaneous excitatory postsynaptic currents (sEPSC’s). These morphological and functional alterations in the cortical inhibitory system characterize the Lis1/sLis1 mouse as a model of mild lissencephaly, showing a phenotype less drastic than the typical phenotype attributed to classical lissencephaly. Therefore, the results described in the present manuscript corroborate the idea that mutations in some regions of the Lis1 gene can produce phenotypes more similar to those typically described in schizophrenic and autistic patients and animal models.
Collapse
Affiliation(s)
- E Domínguez-Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - L Valdés-Sánchez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - S Canals
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - O Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - A Pombero
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - R García-López
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - A Estirado
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - D Pastor
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - E Geijo-Barrientos
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - S Martínez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain.,Centro de Investigación Biomédica en Red en Salud Mental CIBERSAM, Madrid, Spain
| |
Collapse
|
44
|
Liddle PF, Liddle EB. Imprecise Predictive Coding Is at the Core of Classical Schizophrenia. Front Hum Neurosci 2022; 16:818711. [PMID: 35308615 PMCID: PMC8928728 DOI: 10.3389/fnhum.2022.818711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/14/2022] [Indexed: 12/23/2022] Open
Abstract
Current diagnostic criteria for schizophrenia place emphasis on delusions and hallucinations, whereas the classical descriptions of schizophrenia by Kraepelin and Bleuler emphasized disorganization and impoverishment of mental activity. Despite the availability of antipsychotic medication for treating delusions and hallucinations, many patients continue to experience persisting disability. Improving treatment requires a better understanding of the processes leading to persisting disability. We recently introduced the term classical schizophrenia to describe cases with disorganized and impoverished mental activity, cognitive impairment and predisposition to persisting disability. Recent evidence reveals that a polygenic score indicating risk for schizophrenia predicts severity of the features of classical schizophrenia: disorganization, and to a lesser extent, impoverishment of mental activity and cognitive impairment. Current understanding of brain function attributes a cardinal role to predictive coding: the process of generating models of the world that are successively updated in light of confirmation or contradiction by subsequent sensory information. It has been proposed that abnormalities of these predictive processes account for delusions and hallucinations. Here we examine the evidence provided by electrophysiology and fMRI indicating that imprecise predictive coding is the core pathological process in classical schizophrenia, accounting for disorganization, psychomotor poverty and cognitive impairment. Functional imaging reveals aberrant brain activity at network hubs engaged during encoding of predictions. We discuss the possibility that frequent prediction errors might promote excess release of the neurotransmitter, dopamine, thereby accounting for the occurrence of episodes of florid psychotic symptoms including delusions and hallucinations in classical schizophrenia. While the predictive coding hypotheses partially accounts for the time-course of classical schizophrenia, the overall body of evidence indicates that environmental factors also contribute. We discuss the evidence that chronic inflammation is a mechanism that might link diverse genetic and environmental etiological factors, and contribute to the proposed imprecision of predictive coding.
Collapse
Affiliation(s)
- Peter F. Liddle
- Centre for Translational Neuroimaging for Mental Health, School of Medicine, Institute of Mental Health, University of Nottingham, Nottingham, United Kingdom
| | | |
Collapse
|
45
|
Lacaille H, Vacher CM, Penn AA. Preterm Birth Alters the Maturation of the GABAergic System in the Human Prefrontal Cortex. Front Mol Neurosci 2022; 14:827370. [PMID: 35185465 PMCID: PMC8852329 DOI: 10.3389/fnmol.2021.827370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
Developmental changes in GABAergic and glutamatergic systems during frontal lobe development have been hypothesized to play a key role in neurodevelopmental disorders seen in children born very preterm or at/with low birth weight, but the associated cellular changes have not yet been identified. Here we studied the molecular development of the GABAergic system specifically in the dorsolateral prefrontal cortex, a region that has been implicated in neurodevelopmental and psychiatric disorders. The maturation state of the GABAergic system in this region was assessed in human post-mortem brain samples, from term infants ranging in age from 0 to 8 months (n = 17 male, 9 female). Gene expression was measured for 47 GABAergic genes and used to calculate a maturation index. This maturation index was significantly more dynamic in male than female infants. To evaluate the impact of premature birth on the GABAergic system development, samples from 1-month-old term (n = 9 male, 4 female) and 1-month corrected-age very preterm (n = 8 male, 6 female) infants, were compared using the same gene list and methodology. The maturation index for the GABAergic system was significantly lower (−50%, p < 0.05) in male preterm infants, with major alterations in genes linked to GABAergic function in astrocytes, suggesting astrocytic GABAergic developmental changes as a new cellular mechanism underlying preterm brain injury.
Collapse
|
46
|
Seney ML, Glausier J, Sibille E. Large-Scale Transcriptomics Studies Provide Insight Into Sex Differences in Depression. Biol Psychiatry 2022; 91:14-24. [PMID: 33648716 PMCID: PMC8263802 DOI: 10.1016/j.biopsych.2020.12.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a leading cause of disability, affecting more than 300 million people worldwide. We first review the well-known sex difference in incidence of MDD, with women being twice as likely to be diagnosed as men, and briefly summarize how the impact of MDD varies between men and women, with sex differences in symptoms, severity, and antidepressant drug response. We then attempt to deconstruct the biological bases for MDD and discuss implications for sex differences research. Next, we review findings from human postmortem studies, both from selected candidate gene studies and from well-powered, unbiased transcriptomics studies, which suggest distinct, and possibly opposite, molecular changes in the brains of depressed men and women. We then discuss inherent challenges of research on the human postmortem brain and suggest paths forward that rely on thoughtful cohort design. Although studies indicate that circulating gonadal hormones might underlie the observed sex differences in MDD, we discuss how additional sex-specific factors, such as genetic sex and developmental exposure to gonadal hormones, may also contribute to altered vulnerability, and we highlight various nuances that we believe should be considered when determining mechanisms underlying observed sex differences. Altogether, this review highlights not only how various sex-specific factors might influence susceptibility or resilience to depression, but also how those sex-specific factors might result in divergent pathology in men and women.
Collapse
Affiliation(s)
- Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania.
| | - Jill Glausier
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Yan Z, Rein B. Mechanisms of synaptic transmission dysregulation in the prefrontal cortex: pathophysiological implications. Mol Psychiatry 2022; 27:445-465. [PMID: 33875802 PMCID: PMC8523584 DOI: 10.1038/s41380-021-01092-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/13/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
The prefrontal cortex (PFC) serves as the chief executive officer of the brain, controlling the highest level cognitive and emotional processes. Its local circuits among glutamatergic principal neurons and GABAergic interneurons, as well as its long-range connections with other brain regions, have been functionally linked to specific behaviors, ranging from working memory to reward seeking. The efficacy of synaptic signaling in the PFC network is profundedly influenced by monoaminergic inputs via the activation of dopamine, adrenergic, or serotonin receptors. Stress hormones and neuropeptides also exert complex effects on the synaptic structure and function of PFC neurons. Dysregulation of PFC synaptic transmission is strongly linked to social deficits, affective disturbance, and memory loss in brain disorders, including autism, schizophrenia, depression, and Alzheimer's disease. Critical neural circuits, biological pathways, and molecular players that go awry in these mental illnesses have been revealed by integrated electrophysiological, optogenetic, biochemical, and transcriptomic studies of PFC. Novel epigenetic mechanism-based strategies are proposed as potential avenues of therapeutic intervention for PFC-involved diseases. This review provides an overview of PFC network organization and synaptic modulation, as well as the mechanisms linking PFC dysfunction to the pathophysiology of neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. Insights from the preclinical studies offer the potential for discovering new medical treatments for human patients with these brain disorders.
Collapse
Affiliation(s)
- Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | | |
Collapse
|
48
|
Bukina ES, Kondratyev NV, Kozin SV, Golimbet VE, Artyuhov AS, Dashinimaev EB. SLC6A1 and Neuropsychiatric Diseases: The Role of Mutations and Prospects for Treatment with Genome Editing Systems. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421040048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Atefimanash P, Pourhamzeh M, Susanabadi A, Arabi M, Jamali-Raeufy N, Mehrabi S. Hippocampal chloride transporter KCC2 contributes to excitatory GABA dysregulation in the developmental rat model of schizophrenia. J Chem Neuroanat 2021; 118:102040. [PMID: 34695562 DOI: 10.1016/j.jchemneu.2021.102040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
Recent studies have revealed an altered expression of NKCC1 and KCC2 in prefrontal cortex (PFC) and hippocampus of schizophrenic patients. Despite extensive considerations, the alteration of NKCC1 and KCC2 co-transporters at different stages of development has not been fully studied. Therefore, we evaluated the expression of these transporters in PFC and hippocampus at time points of four, eight, and twelve weeks in post-weaning social isolation rearing rat model. For this purpose, 23-25 days-old rats were classified into social- or isolation-reared groups. The levels of NKCC1 and KCC2 mRNA expression were evaluated at hippocampus or PFC regions at the time-points of four, eight, and twelve weeks following housing. Post-weaning isolation rearing decreased the hippocampal KCC2 mRNA expression level, but does not affect the NKCC1 mRNA expression. However, no significant difference was observed in the PFC mRNA levels of NKCC1 and KCC2 in the isolation-reared group compared to the socially-reared group during the course of modeling. Further, we assessed the therapeutic effect of selective NKCC1 inhibitor bumetanide (10 mg/kg), on improvement of prepulse inhibition (PPI) test on twelve weeks isolation-reared rats. Intraperitoneal administration of bumetanide (10 mg/kg) did not exert beneficial effects on PPI deficit. Our findings show that isolation rearing reduces hippocampal KCC2 expression level and may underlie hippocampal GABA excitatory. In addition, 10 mg/kg bumetanide is not effective in improving the reduced PPI of twelve weeks isolation-reared rats. Collectively, our findings show that hippocampal chloride transporter KCC2 contributes to excitatory GABA dysregulation in the developmental rat model of schizophrenia.
Collapse
Affiliation(s)
- Pezhman Atefimanash
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Pourhamzeh
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Susanabadi
- Department of Anesthesia and pain medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mehrnoosh Arabi
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Radiology and Medical Physics, Faculty of Paramedicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Nida Jamali-Raeufy
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Soraya Mehrabi
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran.
| |
Collapse
|
50
|
Förster A, Model V, Gos T, Frodl T, Schiltz K, Dobrowolny H, Meyer-Lotz G, Guest PC, Mawrin C, Bernstein HG, Bogerts B, Schlaaff K, Steiner J. Reduced GABAergic neuropil and interneuron profiles in schizophrenia: Complementary analysis of disease course-related differences. J Psychiatr Res 2021; 145:50-59. [PMID: 34864489 DOI: 10.1016/j.jpsychires.2021.11.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND GABAergic interneuron dysfunction has been implicated in the pathophysiology of schizophrenia. Expression of glutamic acid decarboxylase (GAD), a key enzyme in GABA synthesis, may also be altered. Here, we have simultaneously evaluated GAD-immunoreactive (GAD-ir) neuropil and cell profiles in schizophrenia-relevant brain regions, and analysed disease-course related differences. METHODS GAD65/67 immunoreactivity was quantified in specific brain regions for profiles of fibres and cell bodies of interneurons by automated digital image analysis in post-mortem brains of 16 schizophrenia patients from paranoid (n = 10) and residual (n = 6) diagnostic subgroups and 16 matched controls. Regions of interest were superior temporal gyrus (STG) layers III and V, mediodorsal (MD) and laterodorsal (LD) thalamus, and hippocampal CA1 and dentate gyrus (DG) regions. RESULTS A reduction in GAD-ir neuropil profiles (p < 0.001), particularly in STG layer V (p = 0.012) and MD (p = 0.001), paralleled decreased GAD-ir cell profiles (p = 0.029) in schizophrenia patients compared to controls. Paranoid schizophrenia patients had lower GAD-ir neuron cell profiles in STG layers III (p = 0.007) and V (p = 0.001), MD (p = 0.002), CA1 (p = 0.001) and DG (p = 0.043) than residual patients. There was no difference in GAD-ir neuropil profiles between paranoid and residual subgroups (p = 0.369). CONCLUSIONS These results support the hypothesis of GABAergic dysfunction in schizophrenia. They show a more prominent reduction of GAD-ir interneurons in paranoid versus residual patients, suggestive of more pronounced GABAergic dysfunction in the former. Fully automated analyses of histological sections represent a step towards user-independent assessment of brain structure.
Collapse
Affiliation(s)
- Antonia Förster
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Vera Model
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Tomasz Gos
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Thomas Frodl
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Kolja Schiltz
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Forensic Psychiatry, Mental Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Gabriela Meyer-Lotz
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Christian Mawrin
- Center for Behavioral Brain Sciences, Magdeburg, Germany; Department of Neuropathology, University of Magdeburg, Magdeburg, Germany
| | - Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Bernhard Bogerts
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Salus Institute, Magdeburg, Germany
| | - Konstantin Schlaaff
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Mental Health (DZP), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|